Lignan Glycosides from Neoalsomitra integrifoliola - American

Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, ... of China, and Institute of Materia Medica, Shandong Academy of Medical...
0 downloads 0 Views 105KB Size
784

J. Nat. Prod. 2008, 71, 784–788

Lignan Glycosides from Neoalsomitra integrifoliola Dongmin Su,†,‡ Wenzhao Tang,†,§ Youcai Hu,† Yunbao Liu,† Shishan Yu,*,† Shuanggang Ma,† Jing Qu,† and Dequan Yu† Key Laboratory of BioactiVe Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, People’s Republic of China, Qinghai Institute for Drug Control, Xining 810000, People’s Republic of China, and Institute of Materia Medica, Shandong Academy of Medical Sciences, Jinan 250062, People’s Republic of China ReceiVed October 8, 2007

Five new lignan glycosides, (8R*,7′S*,8′R*)-5,5′-dimethoxy-7-oxolariciresinol 9′-O-β-D-xylopyranoside (1), (7S,8R)dihydrodehydrodiconiferyl alcohol 9-O-β-D-apiofuranosyl-(1f6)-O-β-D-glucopyranoside (2), (7S*,8R*,7′S*,8′R*)-4,4′dimethoxyhuazhongilexin 9-O-β-D-xylopyranoside (4), (7S*,8R*,7′S*,8′R*)-4,4′-dimethoxyhuazhongilexin 9-O-R-Larabinopyranoside (5), and (7S*,8R*,7′S*,8′R*)-huazhongilexin 9-O-(2-feruloyl)-β-D-xylopyranoside (6), together with four known compounds, (7R,8S)-dihydrodehydrodiconiferyl alcohol 9-O-β-D-glucopyranoside (3), the 9-O-β-Dxylopyranoside of icariol A2 (7), huazhongilexin (8), and nudiposide (9), were isolated from the leaves of Neoalsomitra integrifoliola. Their structures were determined by spectroscopic analysis and chemical methods. Compounds 2, 3, 8, and 9 showed weak anti-inflammatory activities. Neoalsomitra integrifoliola (Cogn.) Hutch (Cucurbitaceae) is a toxic shrub distributed in the south and southeast of China. It was reported that the stems, leaves, and fruits of N. integrifoliola were toxic and might cause severe diarrhea or tetany.1 To date, 32 tetracyclic triterpenoids and their saponins were isolated from the bark of the plant.2–5 As part of an ongoing search for active compounds from toxic herbs, we found that the 95% alcohol extract, the n-BuOH-soluble fraction, and its subfraction Fr 3 of the leaves showed weak and moderate anti-inflammatory activities through evaluating mouse ear edema induced by croton oil in ViVo, with inhibitory ratios of 30.3, 45.3, and 49.6%, respectively. Bioassay-guided fractionation of the active fraction led to the isolation of five new lignan glycosides (1, 2, 4-6) and four known compounds, (7R,8S)-dihydrodehydrodiconiferyl alcohol 9-O-β-D-glucopyranoside (3), the 9-O-β-Dxylopyranoside of icariol A2 (7), huazhongilexin (8), and nudiposide (9). Compounds 2, 3, 8, and 9 showed marginal anti-inflammatory activities. Compounds 3, 7, 8, and 9 were isolated from this genus for the first time.

Results and Discussion Compound 1 was obtained as white, amorphous powder. The molecular formula C27H34O13 was determined by HRESIMS indicating 11 degrees of unsaturation. The IR spectrum showed characteristic absorptions for a hydroxy group (3442 cm-1), a carbonyl group (1662 cm-1), and aromatic rings (1603, 1514 cm-1). This observation was further supported by 1H and 13C NMR data. The 1H NMR spectrum of 1 (Table 1) showed proton signals of two 1,3,4,5-tetrasubstituted aromatic rings (δH 7.70, s, 2H, H-2, 6; 7.27, s, 2H, H-2′, 6′), two oxymethylene groups (δH 4.66, dd, J ) 8.5, 6.0 Hz, 1H, H-9a; 4.46, dd, J ) 10.5, 3.5 Hz, 1H, H-9′a; 4.39, t-like, J ) 8.5 Hz, 1H, H-9b; 3.91, dd, J ) 10.5, 4.5 Hz, 1H, H-9′b), and three methines (δH 5.38, d, J ) 8.5 Hz, 1H, H-7′; 4.73, m, 1H, H-8; 3.14, m, 1H, H-8′). In the 13C NMR spectrum, carbon signals of two aromatic rings, a carbonyl, two methylenes, and three methines were observed. These data suggested that 1 was a tetrahydrofuranoid-type lignan.6 The carbonyl was placed at C-7 according to the HMBC correlations (Figure 1). The methoxy * To whom correspondence should be addressed. Tel: 86-10-63165324. Fax: 86-10-63017757. E-mail: [email protected]. † Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College. ‡ Qinghai Institute for Drug Control. § Institute of Materia Medica, Shandong Academy of Medical Sciences.

10.1021/np070565+ CCC: $40.75

groups at δH 3.80 (s, 6H) and 3.83 (s, 6H) in the 1H NMR spectrum were similarly linked to C-3, 5 and 3′, 5′, respectively. From the HMBC correlations, compound 1 was revealed to be a 9-O-7′tetrahydrofuran lignan. The coupling constant of H-7′ (J ) 8.5 Hz) indicated a trans orientation between H-7′ and H-8′.7 The NOE correlation between H-8 (δH 4.73, m) and H-7′ (δH 5.38, d, J ) 8.5 Hz) revealed that H-8 and H-8′ were also trans. The existence of a sugar unit was indicated by the observation of an anomeric proton at δH 4.77 (1H, d, J ) 8.0 Hz) and the corresponding anomeric carbon at δC 105.9 in the 1H and 13C NMR spectra, respectively. The 13C NMR data of the sugar unit were consistent with those of xylose, which was confirmed by acid hydrolysis of 1 and co-TLC with an authentic sample.8 The large coupling constant for the anomeric proton indicated a β-configuration for the xylopyranosyl. According to the literature,9 the sugar unit was assigned as D-xylose by GC analysis. The long-range correlation from H-9′ to C-1′′ suggested that the sugar unit was connected at C-9′. On the basis of this evidence, the structure of 1 was elucidated as (8R*,7′S*,8′R*)-5,5′-dimethoxy-7-oxolariciresinol-9′-O-β-D-xylopyranoside.

 2008 American Chemical Society and American Society of Pharmacognosy Published on Web 04/09/2008

Lignan Glycosides from Neoalsomitra integrifoliola

Journal of Natural Products, 2008, Vol. 71, No. 5 785

Table 1. 1H and 13C NMR Data of Compounds 1-3 (500 MHz for 1H NMR and 125 MHz for 13C NMR) 2 (CD3OD)

1 (C5D5N) no.

δC

1 2 3 4 5 6 7 8 9a 9b OMe 1′ 2′ 3′ 4′ 5′ 6′ 7′ 8′ 9′a 9′b OMe

127.7 107.5 148.9 143.6 148.9 107.5 198.4 49.1 70.9

1′′ 2′′ 3′′ 4′′ 5′′a 5′′b

56.3 131.7 105.1 149.2 137.2 149.2 105.1 84.3 53.2 67.8 56.3 Xyl 105.9 74.9 78.6 71.1 67.3

δH (J in Hz) 7.70, br s

7.70, br s 4.73, m 4.66, dd (11.0, 8.5) 4.39, t (8.5) 3.83, s 7.27, br s

7.27, br s 5.38, d (8.5) 3.14, m 4.46, dd (11.5, 3.0) 3.91, dd (11.5, 4.5) 3.80, s

δC

3 (CD3OD) δH (J in Hz)

134.7 110.8 149.0 147.4 116.0 119.7 89.2 53.0 72.5

6.95, d (1.5) 6.70, d (8.0) 6.80, dd (8.0, 1.5) 5.55, d (6.5) 3.56, m 4.00, m 3.81, m 3.77, s

56.4 136.9 118.2 145.2 147.4 129.5 114.1 32.9 35.8 62.2

6.66, br s 2.57, br t (8.0) 1.76, m 3.51, t (6.5)

56.4 136.9 118.2 145.2 147.5 129.7 114.2 32.9 35.8 62.3

3.80, s

56.8

6.73, br s

56.8

δC

Glu 4.77, d (8.0) 4.06, t (8.5) 4.12, t (8.5) 4.21, m 4.29, dd (11.0, 5.5) 3.66, t (11.0)

1′′ 2′′ 3′′ 4′′ 5′′ 6′′a 6′′b

δH (J in Hz)

134.6 110.7 149.1 147.7 116.1 119.8 89.0 53.2 72.5

6.93, d (1.5) 6.71, d (8.0) 6.80, dd (8.0, 1.5) 5.53, d (6.5) 3.56, m 4.15, dd (9.5, 5.5) 3.82, m 3.83, s 6.74, br s

6.66, br s 2.56, br t (7.5) 1.76, m 3.51, t (6.5) 3.86, s Glu

104.2 75.1 76.9 71.7 78.1 68.5

4.28, d (7.5) 3.16, m 3.30a), m 3.22, m 3.35, m 3.92, dd (11.0, 1.5) 3.56, m

110.9 77.9 80.5 74.9

4.96, d (2.5) 3.82, m

1′′ 2′′ 3′′ 4′′ 5′′ 6′′a 6′′b

104.6 75.2 78.3 71.6 78.1 62.8

4.29, d (8.0) 3.17, m 3.26a), m 3.22, m 3.31, m 3.68, t (10.0) 3.59, m

Api 1′′′ 2′′′ 3′′′ 4′′′a 4′′′b 5′′′ a

65.6

3.87, d (9.5) 3.68, d (9.5) 3.47, m

Overlapped with solvent peak.

Figure 1. Key HMBC correlations of 1. Compound 2 showed a molecular formula of C31H42O15 established by HRESIMS. The IR spectrum showed absorption bands for hydroxy groups (3356 cm-1) and aromatic rings (1607, 1519 cm-1). The 1H and 13C NMR spectra of 2 were similar to those of 3,10,11 except for the additional signals for a sugar unit. Acid hydrolysis of 2 afforded glucose and apiose, which were identified by co-TLC with authentic samples. The large coupling constant of the anomeric proton revealed a β-configuration of the glucose, while the D-form was supported by the GC analysis.9 The presence of β-D-apiose was in accord with the 1H and 13C NMR data.12 Comparison of the 13C NMR data of 2 with those of 3 exhibited that the signal of C-6′′ of the glucose shifted downfield from δC 62.8 to 68.5, suggesting that the apiose was linked at C-6′′, which was further confirmed by long-range correlations from H-1′′′ to C-6′′ and from H-6′′ to C-1′′′ in the HMBC spectrum. The absolute configuration of C-7 was determined by CD data. The positive Cotton effect at 286 nm indicated that the absolute configuration for C-7 was S.13,14 Because of the large coupling constant of H-7, the configuration of C-8 was assigned as R. On the basis of the above evidence, the structure of 2 was elucidated as (7S,8R)-

dihydrodehydrodiconiferyl alcohol 9-O-β-D-apiofuranosyl-(1f6)O-β-D-glucopyranoside. The molecular formulas of compounds 4 and 5 were both established as C29H40O13 by HRESIMS. The IR and UV spectra of 4 and 5 showed very close resemblance to those of 7.15 In the 1H and 13C NMR spectra, two more O-methyl groups in their aglycones were clearly observed compared with 7. These two O-methyl groups were linked at C-4 and C-4′ by HMBC data. Thus, the structure of 4 was elucidated as (7S*,8R*,7′S*,8′R*)-4,4′-dimethoxyhuazhongilexin 9-O-β-D-xylopyranoside. The presence of xylose and arabinose in 4 and 5, respectively, was identified by the 13C NMR data and the co-TLC behavior after acid hydrolysis. The arabinose was indicated as the R-anomer by the coupling constant of the anomeric proton (δH 4.14, d, J ) 7.5 Hz). In the HMBC spectrum of 5, correlation was observed from H-1′′ to C-9, which suggested that the arabinosyl unit was attached to C-9 of the aglycone. Thus, the structure of 5 was elucidated as (7S*,8R*,7′S*,8′R*)-4,4′-dimethoxyhuazhongilexin 9-O-R-L-arabinopyranoside. The HRESIMS of compound 6 displayed an [M + Na]+ ion peak at m/z 767.2482, consistent with a molecular formula of C37H44O16. The IR, UV, and 1H NMR spectra of 6 were also similar to those of 7. Besides the characteristic signals attributed to a 2,5diaryl tetrahydrofuranoid-type lignan glycoside, the 1H NMR spectrum of 6 revealed the presence of a trans-feruloyl moiety (δH 7.12, d, J ) 16.0 Hz, 1H, H-8′′′; 6.06, d, J ) 16.0 Hz, 1H, H-7′′′; 6.76, br s, 1H, H-2′′′; 6.69, br d, J ) 8.8 Hz, 2H, H-5′′′, 6′′′; 3.80, s, 3H), which was further proved by the 13C NMR spectrum.16 By comparing the NMR data of 6 with those of 7, it was found that the signal of H-2′′ shifted downfield by +0.4 ppm, while the signals

786 Journal of Natural Products, 2008, Vol. 71, No. 5

Figure 2. Key HMBC correlations of 2.

Figure 3. Key HMBC correlations of 6. of C-1′′ and C-3′′ shifted upfield by -1 and -2.2 ppm, respectively, which suggested that the trans-feruloyl moiety was located at C-2′′. This linkage position was further confirmed by the correlation from H-2′′ (δH 4.70) to C-9′′′ (δC 165.9) in the HMBC spectrum (Figure 3). Thus, the structure of 6 was elucidated as (7S*,8R*,7′S*,8′R*)huazhongilexin 9-O-(2-feruloyl)-β-D-xylopyranoside. The known compounds were identified as (7R,8S)-dihydrodehydrodiconiferyl alcohol 9-O-β-D-glucopyranoside (3),10,14 the 9-Oβ-D-xylopyranoside of icariol A2 (7),15 huazhongilexin (8),17 and nudiposide (9)18 by comparing with reported spectroscopic data. The EtOH extract showed weak anti-inflammatory activity through evaluating mouse ear edema induced by croton oil in ViVo, with an inhibitory ratio of 30.3%. The anti-inflammatory activities of the fractions were evaluated by the same procedure, and the n-BuOH-soluble fraction and its subfraction Fr 3 exhibited moderate activities with inhibitory ratios of 45.3 and 49.6%, respectively. The anti-inflammatory activities of compounds 1, 2, 3, 7, 8, and 9 isolated from subfraction Fr 3 were assessed by measuring the inhibitory ratios of β-glucuronidase release in rat PMNs induced by PAF in Vitro, and the inhibitory ratios were - 3.89 ( 1.3, 29.2 ( 0.8, 31.1 ( 2.5, 4.67 ( 3.9, 18.1 ( 2.4, and 16.6 ( 2.7% at a concentration of 10-5 mol/L, respectively. Ginkgolide B was used as a positive control, with an inhibitory ratio of 78.4 ( 0.7%. These suggested that compounds 2, 3, 8, and 9 showed marginal inhibitory activities of β-glucuronidase release from rat PMNs induced by PAF. Experimental Section General Experimental Procedures. Melting points were determined on an XT4-100x micromelting point apparatus and were uncorrected. Optical rotations data were determined on a PE model 343 digital polarimeter. UV spectra were obtained on a Hitachi UV-240 spectrophotometer. CD spectra were measured on a JASCO-712 polarimeter. IR spectra were recorded on a Nicolet IMPACT 400 FT-IR spectrometer. NMR spectra were obtained on INOVA-500 and Mercury-400 spectrometers. ESIMS were measured on an Agilent 1100 Series LC/ MSD trap mass spectrometer. HRESIMS data were performed on an Autospec Ultima-TOF mass spectrometer. HPLC was carried out on a Shimadzu LC-6AD equipped with an SPD-10A detector. RP C18 columns (YMC-Pack ODS-A Φ 20 × 250 mm, 5 or 10 µm) were employed for preparative purpose. GC was carried out on a TSQ7000 (Finnigan, America) GC-MS instrument. Solvents were analytical or chromatographic grade and purchased from Beijing Chemical Company, Beijing, China. Macroporous resin D101 (26-60 mesh, Tianjin Haiguang Chemistry Company, Tianjin, China), Sephadex LH-20 (Pharmacia), and ODS (Merck, 20-45, 45-70 µm) were used for CC. Si gel 60 F-254 and HTLC (Qingdao Marine Chemical Factory) were used for TLC. Chromatograms were visualized under UV light or by spraying with 10% H2SO4/EtOH followed by heating.

Su et al. Plant Material. The leaves of N. integrifoliola were collected from Yunnan Province of China in July 2004 and identified by Prof. Lin Ma (Department of Natural Products Chemistry, Institute of Materia Medica, Chinese Academy of Medical Sciences). A voucher specimen (No. 20040361) was deposited in the Herbarium of the Department of Medicinal Plants, Institute of Materia Medica, Chinese Academy of Medical Sciences. Extraction and Isolation. The dried and crushed leaves (4.7 kg) of N. integrifoliola were extracted with 95% EtOH (3 × 30 L) under reflux for 2, 1, and 1 h, respectively. The extracts were combined and concentrated under reduced pressure to yield a dark brown syrup (699 g, 14.9% yield from dried leaves). The EtOH extract showed weak anti-inflammatory activity through evaluating mouse ear edema induced by croton oil in ViVo, with an inhibitory ratio of 30.3%. The EtOH extract was dissolved in H2O (5000 mL) and then partitioned with petroleum ether (3500, 3000, 2000 mL), EtOAc (3500, 3000, 2000 mL), and n-BuOH (3500, 3000, 2000 mL), successively. The n-BuOH-soluble fraction showed moderate anti-inflammatory activity with an inhibitory ratio of 45.3%. This active fraction (120 g) was passed over a D-101 macroporous resin column (2.5 kg) eluting with a gradient of aqueous EtOH (0, 20, 40, 60, 80, 100%, v/v) to yield six fractions (Fr 1-6). The anti-inflammatory activities of the fractions were evaluated in the same procedure, and Fr 3 exhibited moderate activity with an inhibitory ratio of 49.6%. Thus, Fr 3 (27.8 g) was further fractionationed on Sephadex LH-20 (190 g) with 60% MeOH to give nine fractions (Frs 3.1-3.9). Fr 3.3 (3.3 g) was further purified by repeat ODS CC to give compounds 4 (22 mg) and 5 (9 mg). Fr 3.4 (4 g) was subjected to ODS CC (40-75 µm, 200 g) and eluted with 25-70% aqueous MeOH to provide nine fractions (Frs 3.4.1-3.4.9). Compound 7 (60 mg) was obtained from Fr 3.4.3 after repeated ODS CC. Fr 3.4.5 (370 mg) was further purified by repeated ODS CC and preparative HPLC (20% aqueous CH3CN, 4 mL/min, tR ) 45.3 min) to give compound 9 (22 mg). Fr 3.4.7 (324 mg) was applied to repeated ODS, Sephadex LH-20 CC, and preparative HPLC (19% aqueous CH3CN, 5 mL/min, tR ) 41.2 min) to give compound 2 (40 mg). Fr 3.5 (1.9 g) was subjected to ODS CC (40-75 µm, 200 g) and eluted with 20-70% aqueous MeOH to provide 14 fractions (Frs 3.5.1-3.5.14). Compound 1 (40 mg) precipitated from Fr 3.5.9. Fr 3.5.4 (56 mg) was purified by preparative HPLC (30% aqueous MeOH, 5 mL/min) to give compound 8 (23 mg, tR ) 49.5 min). Compound 3 (22 mg) was obtained from Fr 3.5.8 by preparative HPLC (19% aqueous CH3CN, 5 mL/min, tR ) 35.5 min). Fr 3.6 (1.6 g) was subjected to ODS CC and preparative HPLC (19% aqueous CH3CN, 5 mL/min) to give compound 6 (25 mg, tR ) 28.1 min). (8R*,7′S*,8′R*)-5,5′-Dimethoxy-7-oxolariciresinol 9′-O-β-D-xylopyranoside (1): white, amorphous powder; [R]20D -77.3 (c 0.14, MeOH); UV (MeOH) λmax (log ) 209 (2.20), 229 (1.02), 302 (0.59) nm; IR νmax 3442, 2899, 1662, 1603, 1514, 1101, 1036 cm-1; 1H and 13 C NMR data, see Table 1; HRESIMS m/z 589.1884 for [M + Na]+ (calcd for C27H34O13Na, 589.1897). (7S,8R)-Dihydrodehydrodiconiferyl alcohol 9-O-β-D-apiofuranosyl-(1f6)-O-β-D-glucopyranoside (2): white, amorphous powder; [R]20D -25.7 (c 0.10, MeOH); UV (MeOH) λmax (log ) 207 (1.84), 233 (0.48), 281 (0.18) nm; CD (MeOH) λmax (∆) 263 (0.01), 270 (1.09), 275 (2.89), 280 (4.62), 290 (6.61), 295 (4.00), 298 (0.97), 303 (-1.78) nm; IR νmax 3356, 2934, 1607, 1519, 1039 cm-1; 1H and 13C NMR data, see Table 1; ESIMS m/z 693 [M + K]+, 677 [M + Na]+, 653 [M - H]-; HRESIMS m/z 677.2371 for [M + Na]+ (calcd for C31H42O15Na, 677.2421). (7S*,8R*,7′S*,8′R*)-4,4′-Dimethoxyhuazhongilexin 9-O-β-D-xylopyranoside (4): white, amorphous powder; [R]20D -23.0 (c 0.11, MeOH); UV (MeOH) λmax (log ) 207 (1.57), 225 (0.16) nm; IR νmax 3387, 2901, 1640, 1592, 1504, 1421, 1235, 1042 cm-1; 1H and 13C NMR data, see Table 2; ESIMS m/z 635 [M + K]+, 619 [M + Na]+, 595 [M - H]-; HRESIMS m/z 619.2345 for [M + Na]+ (calcd for C29H40O13Na, 619.2367). (7S*,8R*,7′S*,8′R*)-4,4′-Dimethoxyhuazhongilexin 9-O-r-L-arabinopyranoside (5): white, amorphous powder; [R]20D +1 (c 0.04, MeOH); UV (MeOH) λmax (log ) 204 (1.34), 223 (0.67) nm; IR νmax 3381, 2903, 1644, 1592, 1504, 1421, 1235, 1042 cm-1; 1H and 13C NMR data, see Table 2; ESIMS m/z 635 [M + K]+, 619 [M + Na]+, 595 [M - H]-; HRESIMS m/z 619.2328 for [M + Na]+ (calcd for C29H40O13Na, 619.2367).

Lignan Glycosides from Neoalsomitra integrifoliola

Journal of Natural Products, 2008, Vol. 71, No. 5 787

Table 2. 1H and 13C NMR Data of Compounds 4-6 4 (CD3OD)c no.

δC

1 2, 6 3, 5 4 OMe-3, 5 7 8 9a 9b 1′ 2′, 6′ 3′, 5′ 4′ OMe-3′, 5′ OMe-4, 4′ 7′ 8′ 9′a 9′b Xyl 1′′ 2′′ 3′′ 4′′ 5′′a 5′′b

139.8 104.4 154.6 138.5 56.6 84.3 52.2 68.9 139.9 104.6 154.6 138.4 56.7 61.1 84.0 54.3 61.0 105.2 75.0 78.0 71.2 67.1

δH (J in Hz) 6.71, s 3.81, s 5.04, d (8.0) 2.34, m (W1/2 ) 13.3) 3.88, dd (11.5, 3.5) 3.67, dd (11.5, 3.5) 6.73, s 3.81, s 3.70, s 4.99, d (8.0) 2.34, m (W1/ 2) 13.3) 3.73, m 3.60, dd (11.5, 2.0) 4.17, d (7.5) 3.15, t (8.0) 3.24a 3.43, m 3.79b 3.12, dd (13.5, 2.50)

5 (CD3OD)c δC 139.8 104.5 154.6 138.5 56.7 84.3 52.2 68.7 139.9 104.6 154.6 138.4 56.7 61.1 84.1 54.3 61.1 105.1 72.5 74.4 69.7 67.2

δH (J in Hz) 6.71, s 3.81, s 5.07, d (8.0) 2.36, m (W1/2 ) 12.7) 3.99, dd (10.5, 4.0) 3.67, dd (10.5, 4.0) 6.75, s 3.81, s 3.70, s 4.99, d (8.0) 2.36, m (W1/ 2) 12.7) 3.73, dd (11.5, 3.0) 3.60, dd (11.5, 3.0) 4.14, d (7.5) 3.50, m 3.45, dd (9.5, 3.0) 3.75, m 3.79b 3.50, m

6 (DMSO-d6)c δC

δC

δH (J in Hz)

132.8 103.1 147.7 134.5 55.8 81.7 49.3 67.1

6.55, s 3.76, s 4.85, d (8.5) 2.19, ddt (8.5, 9.5, 4.5) 3.72b 3.46, m

132.7 103.8 147.8 134.7 55.8

3.76, s 4.76, d (8.5) 2.02, ddt (8.5, 9.5, 4.5) 3.32, dd (11.5, 3.5) 3.16, m

101.7 73.4 74.0 69.8 65.9

4.42, d (8.0) 4.70, t (8.5) 3.37, t (8.5) 3.42, m 3.79b 3.16, m 124.4 110.6 147.8 148.1 115.5 123.1 113.5 145.0 165.9 55.5

127.1 111.6 149.3 151.2 116.5 123.8 114.8 147.2 168.1 56.4

6.66, br d (8.5) 6.77, br d (8.5) 6.23, d (16.0) 7.19, d (16.0) 3.75, s

Overlapped with solvent peak. b Overlapped with -OMe peak. c 500 MHz for 1H NMR and 125 MHz for 100 MHz for 13C NMR.

83.9 52.2 58.7 104.3 75.0 75.8 71.4 67.2

7.01, br s

a

136.0 104.9 149.2 134.2 56.7 84.1 50.7 67.3 136.0 104.4 149.3 134.0 56.7

6.58, s

81.4 51.7 58.1

feruyl 1′′′ 2′′′ 3′′′ 4′′′ 5′′′ 6′′′ 7′′′ 8′′′ 9′′′ OMe

(7S*,8R*,7′S*,8′R*)-Huazhongilexin 9-O-(2-feruloyl)-β-D-xylopyranoside (6): pale yellow, amorphous powder; [R]20D -33.3 (c 0.20, MeOH); UV (MeOH) λmax (log ) 208 (2.21), 238 (0.69), 325 (0.47) nm; IR νmax 3405, 2936, 2844, 1701, 1602, 1515, 1325, 1113, 830 cm-1; 1H and 13C NMR data, see Table 2; ESIMS m/z 767 [M + Na]+, 743 [M - H]-; HRESIMS m/z 767.2482 for [M + Na]+ (calcd for C37H44O16Na, 767.2527). Acid Hydrolysis of Compounds 1-5 and 7. Compounds 1 (8 mg) and 2 (11 mg) were dissolved in 5% HCl (50 mL) and heated at 90 °C for 8 h. The mixture was evaporated under vacuum to 20 mL and then extracted with EtOAc three times (25, 25, 20 mL). After addition of H2O, the acidic solution was evaporated again and then dried in Vacuo to furnish a monosaccharide residue. From the residue, xylose, glucose, and apiose were detected by co-TLC [CHCl3-MeOH-HOAc-H2O (25:12:2:2)] with authentic samples. The Rf values for the above sugars were 0.43, 0.20, and 0.66, respectively. Compounds 3-5 and 7 were individually hydrolyzed with HCl vapor on an HPTLC precoated plate (80 °C water bath for 40 min) followed by developing with CHCl3-MeOH-HOAc-H2O (25:12:2:2).9 The sugars were visualized by spraying with phenylamine-ortho-benzenedicarboxylic acid reagent followed by heating, and compared with authentic samples (Rf of D-glucose, D-xylose, L-arabinose: 0.21, 0.44, 0.39). Absolute Configuration of the Sugars. The absolute configuration of the xylose and glucose was determined according to a reported procedure.9 An authentic sugar sample (3 mg) was trimethylsilylated, and the derivative was analyzed by GC-MS. The monosaccharide residues obtained from the acid hydrolysis of compounds 1 and 2 were treated as the authentic sugar samples following the same procedures, and the retention times of sugar derivatives were compared with those of the authentic samples (D-xylose 17.67 min, D-glucose 19.57 min).

6 (CD3OD)d

13C

δH (J in Hz) 6.63, s 3.78, s 5.04, d (8.0) 2.23, m 3.90, br d (10.0) 3.76b 6.61, s 3.83, s 4.84, d (8.5) 2.23. m 3.81b 3.46, dd (12.4, 3.0) 4.36, d (7.6) 3.50, t (8.5) 3.22, m 3.58, m 3.83b 3.22, m 6.76, br s 6.69, br d (8.8) 6.69, br d (8.8) 6.06, d (16.0) 7.12, d (16.0) 3.80, s

NMR. d 400 MHz for 1H NMR and

Anti-inflammatory Bioassays. The anti-inflammatory activities of the EtOH extract and the fractions were measured through evaluating mouse ear edema induced by croton oil in ViVo.19 According to the reported procedures,20 the anti-inflammatory activities of compounds 2, 3, 7, 8, and 9 were assessed by measuring the inhibition of the PAFinduced release of β-glucuronidase from rat polymorphonuclear leukocytes (PMNs) in Vitro. The absorbance was read at 550 nm, and the inhibitory ratio was calculated. Ginkgolide B was used as reference compound. Acknowledgment. The research project was supported by the National Science Fund for Distinguished Young Scholars (No. 30625040), the Key Project of Natural Science Foundation of China (No. 20432030), “973” Project (No. 2004CB518906), and PCSIRT (No. IRT0514). We are grateful to the Department of Instrumental Analysis of the Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, for the measurements of UV, IR, NMR, and FABMS spectra, and the Department of Pharmacology of our Institute for measuring antiinflammatory activities. References and Notes (1) Cheng, J. S.; Zhen, S. Chinese Virose Plant, 1st ed.; Science Press: Beijing, 1987; p 205. (2) Fujita, S.; Kasai, R.; Ohtani, K.; Yamasaki, K.; Chiu, M. H.; Nie, R. L.; Tanaka, O. Phytochemistry 1995, 38, 465–472. (3) Fujita, S.; Kasai, R.; Ohtani, K.; Yamasaki, K.; Chiu, M. H.; Nie, R. L.; Tanaka, O. Phytochemistry 1995, 39, 591–602. (4) Chiu, M. H.; Nie, R. L.; Hiromichi, N.; Akira, I.; Zhou, J.; Akinori, S. Phytochemistry 1992, 31, 2451–2453.

788 Journal of Natural Products, 2008, Vol. 71, No. 5 (5) Chiu, M. H.; Nie, R. L.; Li, Z. R.; Kasai, R.; Zhou, J. Acta Bot. Yunnan. 1992, 14, 442–444. (6) Kinjo, J.; Higuchi, H.; Fukui, K.; Nohara, T. Chem. Pharm. Bull. 1991, 39, 2952–2955. (7) Go´ngora, L.; Ma´n˜ez, S.; Giner, R. M.; Recio, M. C.; Gray, A. I.; Rı´os, J. L. Phytochemistry 2002, 59, 857–860. (8) Zhao, P. P.; Li, B. M.; He, L. Y. Acta Pharm. Sin. 1987, 22, 70–74. (9) Kinjo, J.; Araki, K.; Fukui, K.; Higuchi, H.; Ikeda, T.; Nohara, T.; Ida, Y.; Takemoto, N.; Miyakoshi, M.; Shoji, J. Chem. Pharm. Bull. 1992, 40, 3269–3273. (10) Takeda, Y.; Mima, C.; Masuda, T.; Hirata, E.; Takuahi, A.; Otsuka, H. Phytochemistry 1998, 49, 2137–2139. (11) Baderschneider, B.; Winterhalter, P. J. Agric. Food Chem. 2001, 49, 2788–2798. (12) Li, Y.; Zhang, D. M.; Li, J. B.; Yu, S. S.; Li, Y.; Luo, Y. M. J. Nat. Prod. 2006, 69, 616–620.

Su et al. (13) Miyase, T.; Ueno, A.; Takizawa, N.; Kobayashi, H.; Oguchi, H. Phytochemistry 1989, 28, 3483–3485. (14) Otsuka, H.; Hirata, E.; Shinzato, T.; Takeda, Y. Chem. Pharm. Bull. 2000, 48, 1084–1086. (15) Takara, K.; Matsui, D.; Wada, K.; Ichiba, T.; Chinen, I.; Nakasone, Y. Biosci. Biotechnol. Biochem. 2003, 67, 376–379. (16) Hosny, M.; Rosazza, J. P. N. J. Nat. Prod. 1997, 60, 219–222. (17) Lin, L. D.; Qin, G. W.; Xu, R. S.; Tian, Z. Y.; Lv, Y.; Zheng, Q. T. Acta Chim. Sin. 1995, 3, 98–101. (18) Fuchino, H.; Satoh, T.; Tanaka, N. Chem. Pharm. Bull. 1995, 43, 1937– 1942. (19) Manga, H. M.; Brkic, D.; Marie, D. E. P.; Quetin, L. J. J. Ethnopharm. 2004, 92, 209–214. (20) Kang, J.; Chen, R. Y.; Yu, D. Q. Planta Med. 2006, 72, 52–59.

NP070565+